Signal peptidase complex subunit 2 (SPCS2) antibody, also referred to as SPC25 antibody in some literature, is an immunological reagent specifically designed to target and bind to the SPCS2 protein component of the signal peptidase complex (SPC). This complex is responsible for cleaving N-terminal signal sequences from nascent proteins as they are translocated into the lumen of the endoplasmic reticulum . The Spc2 antibody has emerged as an indispensable tool for investigating the structure, function, and interactions of the signal peptidase complex, helping researchers elucidate the mechanisms by which cells process secretory and membrane proteins .
The target of Spc2 antibody, Signal peptidase complex subunit 2 (SPCS2), is an integral membrane protein component of the signal peptidase complex. In eukaryotes, the SPC comprises four evolutionarily conserved membrane subunits (Spc1–3 and Sec11) . SPCS2 has a molecular weight of approximately 18 kDa in yeast and 25 kDa in humans . The protein contains transmembrane domains and a cytosolic C-terminal domain that plays a significant role in substrate recognition .
SPCS2 enhances the enzymatic activity of the signal peptidase complex and facilitates interactions between different components of the translocation site. Recent research has revealed that SPCS2 modulates the properties of the SPC and its surrounding membrane environment, thereby improving the complex's ability to discriminate between signal peptides (SPs) and signal-anchored sequences (SAs) .
The SPCS2 protein structure is well conserved between yeast and human, with both containing similar cytosolic domains that constitute much of the cytosolic part of the signal peptidase complex . This evolutionary conservation underscores the fundamental importance of SPCS2 in protein processing across eukaryotic species.
Spc2 antibodies are frequently employed in Western blotting to detect and quantify SPCS2 protein levels in cellular extracts. This application has been vital in studies examining the expression levels of SPCS2 across different tissues, cell types, and experimental conditions. For instance, researchers have used Western blotting with Spc2 antibodies to assess the steady-state levels of Spc2 and its variants in yeast strains, confirming protein expression at the expected sizes .
Immunohistochemistry (IHC) and immunocytochemistry (ICC) utilizing Spc2 antibody provide valuable insights into the cellular and subcellular localization of SPCS2. These techniques have demonstrated that SPCS2 is primarily localized to the endoplasmic reticulum membrane, consistent with its role in signal peptide processing . The antibody has been validated for IHC applications on paraffin-embedded tissue sections, enabling studies of SPCS2 expression in various tissues and pathological conditions.
Spc2 antibodies have played a crucial role in co-immunoprecipitation experiments aimed at identifying protein-protein interactions. These studies have helped elucidate the interactions between SPCS2 and other components of the signal peptidase complex, as well as potential interactions with substrate proteins and translocon components .
Research utilizing Spc2 antibodies has significantly contributed to our understanding of signal sequence processing mechanisms. A landmark study published in 2024 employed Spc2 antibodies to investigate how the Signal peptidase complex distinguishes signal peptides from signal-anchored sequences . The experiments revealed that SPCS2 modulates substrate recognition and cleavage site identification. Specifically, when SPCS2 was absent or mutated, the discrimination between substrates and identification of cleavage sites by the SPC was compromised .
Investigations using Spc2 antibodies have demonstrated the critical importance of the C-terminal domain of SPCS2 in N-length dependent signal sequence cleavage. Researchers constructed SPCS2 mutants lacking the C-terminal domain (either 58 or 23 residues) and then used antibodies to detect these variants in Western blotting experiments. The results showed that the C-terminal domain of SPCS2 significantly affects the SPC's ability to process signal sequences with varying N-region lengths .
One of the most intriguing findings facilitated by Spc2 antibody research is the role of SPCS2 in modulating membrane thickness. Through a combination of molecular dynamics simulations and experimental validation using Spc2 antibodies, researchers discovered that SPCS2 causes membrane thinning at the center of the SPC. This thinning is approximately 3 Å compared to membranes without SPCS2, allowing the complex to better discriminate between signal peptides with shorter hydrophobic regions and signal-anchored sequences with longer hydrophobic segments .
Protein Variant | Membrane Thickness in TM Window (Å) | Cleavage Efficiency of Short h-region | Cleavage Efficiency of Long h-region |
---|---|---|---|
Wild-type SPCS2 | ~21 | High | Low |
No SPCS2 | ~24 | High | High |
SPCS2 Y79A,S83A | ~22 | High | Intermediate |
Note: h-region refers to the hydrophobic region of signal sequences
Research using Spc2 antibodies has extended beyond basic cellular mechanisms to include virological studies. For example, investigators have used these antibodies to examine the role of SPCS2 in viral assembly. In the case of Hepatitis C virus (HCV), studies suggest that signal peptidase complex components, including SPCS2, may interact with viral proteins to facilitate viral particle assembly .
Spc2 antibodies are typically generated through immunization of host animals (commonly rabbits) with purified recombinant SPCS2 protein or synthetic peptides corresponding to specific regions of SPCS2. For instance, Abcam's Anti-SPCS2/SPC25 antibody (ab121395) was produced using a recombinant fragment corresponding to human SPCS2 amino acids 1-100 as the immunogen . Following immunization, antibodies are harvested from serum and purified using antigen affinity methods to enhance specificity.
Commercial Spc2 antibodies undergo rigorous validation to ensure specificity and performance across various applications. Validation typically includes Western blotting against cell lines known to express SPCS2, such as RT-4, U-251 MG, and human liver samples . Additional validation may involve immunocytochemistry/immunofluorescence on cell lines like U-2 OS, with antibody concentrations optimized for each specific application .
Application | Cell/Tissue Type | Antibody Concentration | Detection Method | Result |
---|---|---|---|---|
Western Blot | RT-4 | 1/250 dilution | ECL | Detected band at ~25 kDa |
Western Blot | U-251 MG | 1/250 dilution | ECL | Detected band at ~25 kDa |
Western Blot | Human liver | 1/250 dilution | ECL | Detected band at ~25 kDa |
ICC/IF | U-2 OS | 2 μg/mL | Fluorescence | Positive staining in ER pattern |
Note: ECL = Enhanced Chemiluminescence; ICC/IF = Immunocytochemistry/Immunofluorescence; ER = Endoplasmic Reticulum
Spc2 antibodies have been instrumental in genetic studies investigating the effects of SPCS2 mutations or deletions. In a notable study, researchers created yeast strains with various SPCS2 mutations and used Spc2 antibodies to confirm protein expression and assess the impact on signal peptidase complex function .
To verify the expression of SPCS2 mutants in yeast cells, researchers utilized hemagglutinin (HA) tag fusion and then performed Western blotting with anti-HA antibodies. Additionally, they employed quantitative mass spectrometry to compare the relative protein abundances of signal peptidase complex components between wild-type and mutant strains . This approach enabled a comprehensive analysis of how SPCS2 mutations affect the composition and function of the signal peptidase complex.
While the primary applications of Spc2 antibody have been in basic research elucidating the mechanisms of protein processing, emerging evidence suggests potential relevance to disease research. Signal peptide processing defects have been implicated in various pathological conditions, including certain neurodegenerative disorders and congenital disorders of glycosylation .
For instance, in a study investigating prohormone processing in pancreatic islets, researchers used an antibody against the C-terminus of SPC2 to examine the effects of SPC2 deficiency on islet morphology and function. The results showed that SPC2 deficiency led to impaired prohormone processing and altered pancreatic islet morphology, with implications for understanding certain metabolic disorders .
The continuous development and refinement of Spc2 antibodies promise to advance our understanding of signal peptide processing in several key areas:
Further research using Spc2 antibodies may uncover connections between aberrant signal peptide processing and various pathological conditions. This could lead to the development of diagnostic tools or therapeutic strategies targeting the signal peptidase complex .
Spc2 antibodies could facilitate high-throughput screening assays for compounds that modulate signal peptidase activity, potentially leading to the development of novel therapeutics targeting protein processing pathways.
KEGG: spo:SPAC1071.04c
STRING: 4896.SPAC1071.04c.1
SPC2 (SPCS2 in humans, Spc2 in yeast) is a subunit of the signal peptidase complex (SPC) that catalyzes the cleavage of N-terminal signal sequences from nascent proteins as they are translocated into the lumen of the endoplasmic reticulum . In yeast, Spc2 has been shown to modulate substrate- and cleavage site-selection, playing a critical role in discriminating between signal peptides (SPs) and signal-anchored (SAs) sequences .
The SPC comprises four evolutionarily conserved membrane subunits (Spc1–3 and Sec11), with Spc2 enhancing the enzymatic activity of the complex and facilitating interactions between different components of the translocation site . Importantly, Spc2 interacts with the β subunit of the Sec61 translocon in yeast and mammals, mediating transient interactions between the SPC and the Sec61 translocon .
This is a critical distinction researchers must understand to avoid experimental confusion. SPC2 (SPCS2/SPC25) antibodies target the signal peptidase complex subunit 2, which is involved in protein translocation and signal peptide cleavage at the endoplasmic reticulum .
In contrast, SCP2 (Sterol carrier protein-2) antibodies target a completely different protein involved in intracellular lipid transport and metabolism . SCP2 is also called nonspecific lipid-transfer protein and plays no role in signal peptide processing . When ordering or using these antibodies, researchers should carefully verify the correct target to prevent experimental errors.
The human SPCS2/SPC25 protein has a predicted band size that corresponds to its role as the ~25 kDa subunit of the signal peptidase complex . This is consistent with its alternative name "Microsomal signal peptidase 25 kDa subunit." When conducting Western blot experiments, researchers should expect to observe bands in this range, though post-translational modifications might cause slight variations in the observed molecular weight.
Based on available data, antibodies targeting human SPCS2/SPC25 have been validated for use in human cell lines, particularly K562 cells (human chronic myelogenous leukemia cell line) . When studying the yeast Spc2 ortholog, researchers have successfully used specific antibodies in Saccharomyces cerevisiae systems .
For SCP2 antibodies (distinct from SPC2), validation has been performed in multiple human cell lines including HEK-293T, HEK-293, K-562, HepG2, and mouse NIH/3T3 cells . Researchers should conduct preliminary validation experiments when applying these antibodies to new cell types or tissue systems.
Anti-SPCS2/SPC25 antibodies have been validated for multiple experimental applications:
Western Blot (WB): Effective for detecting SPCS2 protein expression levels
Immunohistochemistry-Paraffin (IHC-P): Suitable for tissue section analysis
Immunocytochemistry/Immunofluorescence (ICC/IF): For cellular localization studies
For comparison, SCP2 antibodies (targeting the different lipid transport protein) have been validated for:
Application | Recommended Dilution |
---|---|
Western Blot (WB) | 1:1000-1:4000 |
Immunoprecipitation (IP) | 0.5-4.0 μg for 1.0-3.0 mg of total protein lysate |
Immunofluorescence (IF)/ICC | 1:20-1:200 |
Flow Cytometry (FC) (INTRA) | 0.40 μg per 10^6 cells in a 100 μl suspension |
Researchers should note that optimal dilutions may vary between experimental systems and should be determined empirically .
SPC2 antibodies can be instrumental in investigating the structure and function of the signal peptidase complex. One advanced approach involves combining immunoprecipitation with SPC2 antibodies followed by mass spectrometry to identify interaction partners within the complex and associated proteins.
In yeast studies, researchers have used antibodies against Spc2 and other SPC components (like Spc3) to investigate how mutations or deletions affect complex formation. For instance, western blotting with anti-HA tagged Spc3 was used to assess Spc3 stability in Spc2 mutant cells . Similar approaches can be applied in mammalian systems using SPCS2 antibodies.
Co-immunoprecipitation experiments can reveal how SPCS2/Spc2 mediates interactions with the Sec61 translocon, providing insights into the molecular mechanisms underlying protein translocation and signal peptide processing.
When interpreting results from Spc2 deletion or knockdown experiments, researchers should consider several factors:
Substrate specificity changes: In yeast Spc2 deletion strains, cleavage efficiency is altered in a substrate-dependent manner. Specifically, processing of signal sequences with longer hydrophobic regions (h-regions) is increased in the absence of Spc2, while processing of sequences with shorter h-regions may be decreased .
Membrane environment effects: Molecular dynamics simulations suggest that Spc2 affects membrane thickness around the SPC complex. Without Spc2, the membrane is approximately 3Å thicker in the transmembrane window compared to wild-type SPC . This physical change may explain alterations in substrate specificity.
Compensatory mechanisms: While Spc2 contributes to SPC function, its absence does not completely abolish signal peptide processing. In yeast, signal peptides of secretory precursors can still be efficiently processed in the absence of Spc2 . This suggests compensatory mechanisms that researchers should account for in their experimental design.
Stability of other SPC components: When manipulating Spc2 expression, researchers should verify that other SPC components remain stable. In yeast studies, the expression levels of Sec11, Spc3, and Sbh2 were assessed by quantitative mass spectrometry in Spc2 deletion strains .
Distinguishing between defects in signal peptide processing (SPC function) and protein translocation requires careful experimental design:
Pulse-chase analysis: Researchers can use metabolic labeling followed by immunoprecipitation to track the processing kinetics of secretory proteins. In SPC2 deficiency, precursor forms would accumulate but still enter the ER, whereas translocation defects would prevent ER entry entirely.
Subcellular fractionation: By separating cytosolic and membrane fractions, researchers can determine if unprocessed precursors are associated with membranes (suggesting SPC defects) or remain cytosolic (suggesting translocation defects).
Model substrate analysis: Using engineered substrates with varied signal sequence properties (different n-region lengths, h-region hydrophobicity, etc.) can help distinguish processing from translocation defects. Research shows that Spc2 particularly affects the processing of substrates with specific n-region and h-region characteristics .
Electron microscopy: Ultrastructural analysis of the ER can reveal membrane abnormalities or protein aggregation that might distinguish between processing and translocation defects.
For optimal immunofluorescence results with SPCS2/SPC25 antibodies:
Fixation: Use 4% paraformaldehyde for 15-20 minutes at room temperature to preserve membrane protein structure.
Permeabilization: Since SPCS2 is a membrane protein with domains facing both cytosol and ER lumen, use 0.1-0.2% Triton X-100 for permeabilization.
Blocking: Use 5% normal serum (from the species in which the secondary antibody was raised) with 0.1% BSA to minimize background.
Antibody dilution: Start with a 1:50 to 1:200 dilution range for primary SPCS2 antibodies . Optimize empirically for your specific system.
Co-staining markers: Consider co-staining with ER markers (calnexin, PDI) to confirm localization to the ER membrane.
Controls: Include appropriate negative controls (secondary antibody only, isotype control) and positive controls (cell types known to express SPCS2).
Quantitative assessment of SPCS2 expression can be achieved through several methods:
Western blot densitometry: Use validated SPCS2 antibodies for western blotting, followed by densitometric analysis normalized to appropriate loading controls (GAPDH, β-actin). When comparing across multiple conditions, include a standard curve with known protein amounts.
Quantitative mass spectrometry: For higher precision, researchers can employ stable isotope labeling with amino acids in cell culture (SILAC) or tandem mass tag (TMT) approaches. This has been successfully used to compare protein abundance between wild-type and Spc2 mutant yeast cells .
Flow cytometry: For single-cell quantification, intracellular staining with SPCS2 antibodies can be performed. Based on protocols for similar intracellular proteins, use approximately 0.4 μg antibody per 10^6 cells .
RT-qPCR: While measuring mRNA rather than protein, this can provide complementary data on expression regulation. Validate findings at the protein level due to potential post-transcriptional regulation.
Image analysis of immunofluorescence: Mean fluorescence intensity measurements from confocal microscopy can provide semi-quantitative data on expression levels and subcellular distribution.
While the search results don't provide specific information about SPCS2 in disease states, general principles can be inferred based on its biological function:
As a component of the signal peptidase complex, SPCS2 is involved in the processing of numerous secretory and membrane proteins. Dysregulation of this processing machinery could potentially affect multiple cellular pathways and contribute to various disease states, particularly those involving protein misfolding or secretory pathway defects.
Researchers investigating SPCS2 in disease contexts should consider:
ER stress responses: Defects in signal peptide processing may trigger unfolded protein responses and ER stress, which are implicated in neurodegenerative diseases and diabetes.
Secretory protein processing: Alterations in SPCS2 function could impact the maturation of secreted proteins, potentially affecting hormone processing, immune function, or extracellular matrix formation.
Viral infections: Many viral proteins require signal peptide processing for maturation. SPCS2 dysfunction or targeting could potentially affect viral replication cycles.
SPC2 antibodies can be valuable tools for investigating protein translocation and processing defects in disease models:
Comparative expression analysis: Assess SPCS2 levels in normal versus diseased tissues/cells using immunohistochemistry or western blotting with validated antibodies .
Colocalization studies: Combine SPCS2 antibodies with markers of ER stress (BiP/GRP78, phospho-PERK) to investigate whether signal peptidase complex dysfunction correlates with ER stress responses in disease models.
Protein processing analysis: Use SPCS2 antibodies in pulse-chase experiments to compare the kinetics of signal sequence processing between normal and disease states.
Interaction partner identification: Employ co-immunoprecipitation with SPCS2 antibodies followed by mass spectrometry to identify altered protein interactions in disease states that might contribute to pathology.
Therapeutic target validation: In models where signal peptide processing defects are implicated, SPCS2 antibodies can help validate the signal peptidase complex as a potential therapeutic target.
The discovery that Spc2 affects membrane thickness around the signal peptidase complex opens exciting new research directions:
Lipid-protein interactions: Researchers could investigate how specific lipid compositions affect SPC2 function and signal peptide processing efficiency. This might involve reconstitution experiments with defined lipid compositions or targeted lipidomic analysis of ER membranes in SPC2-manipulated cells.
Nanoscale membrane organization: Advanced imaging techniques such as super-resolution microscopy could be employed to visualize how SPC2 influences the nanoscale organization of the ER membrane and the distribution of other translocation components.
Membrane-targeting therapeutics: Understanding how SPC2 modulates membrane properties could inform the development of therapeutic approaches that target the membrane environment of the signal peptidase complex rather than the protein itself.
Evolutionary adaptations: Comparative analysis of SPC2 sequences across species could reveal adaptations that optimize signal peptide processing in different membrane environments or cellular contexts.
The role of Spc2 in modulating substrate recognition and cleavage site selection has significant implications for synthetic biology:
Engineered secretion systems: Understanding how Spc2 contributes to signal sequence discrimination could enable the design of optimized signal sequences for improved secretion of recombinant proteins in biotechnology applications.
Controllable protein localization: By engineering signal sequences with characteristics that make them more or less dependent on Spc2 for processing, researchers might develop systems for controllable protein localization between different cellular compartments.
Synthetic organelle targeting: Insights from Spc2 research could inform the design of synthetic targeting sequences for novel organelles or membrane compartments in synthetic biology applications.
Biomolecular condensate targeting: Understanding the principles of how Spc2 recognizes specific features of signal sequences (such as the n-region length dependence ) could inform strategies for targeting proteins to biomolecular condensates or other non-membrane-bound compartments.